<<

Survival Analysis Using a 5-Step Stratified Testing and Amalgamation Routine (5-STAR) in Randomized Clinical Trials Devan V. Mehrotra* and Rachel Marceau West and Research Decision Sciences, Merck & Co., Inc., North Wales, PA, USA * E-mail: [email protected]

ABSTRACT Randomized clinical trials are often designed to assess whether a test treatment prolongs survival relative to a control treatment. Increased patient heterogeneity, while desirable for generalizability of results, can weaken the ability of common statistical approaches to detect treatment differences, potentially hampering the regulatory approval of safe and efficacious therapies. A novel solution to this problem is proposed. A list of baseline covariates that have the potential to be prognostic for survival under either treatment is pre-specified in the analysis plan. At the analysis stage, using all observed survival times but blinded to patient-level treatment assignment, ‘noise’ covariates are removed with elastic net Cox regression. The shortened covariate list is used by a conditional inference tree algorithm to segment the heterogeneous trial population into subpopulations of prognostically homogeneous patients (risk strata). After patient-level treatment unblinding, a treatment comparison is done within each formed risk stratum and stratum-level results are combined for overall . The impressive power-boosting performance of our proposed 5-step stratified testing and amalgamation routine (5-STAR), relative to that of the and other common approaches that do not leverage inherently structured patient heterogeneity, is illustrated using a hypothetical and two real datasets along with simulation results. Furthermore, the importance of reporting stratum-level comparative treatment effects (time ratios from accelerated failure time model fits in conjunction with model averaging and, as needed, hazard ratios from Cox proportional hazard model fits) is highlighted as a potential enabler of personalized medicine. A fiveSTAR R package is available at https://github.com/rmarceauwest/fiveSTAR.

KEYWORDS: conditional inference tree, elastic net regression, model averaging, risk stratum, stratified medicine, time ratio

1

1. INTRODUCTION

Consider a typical randomized designed to compare the effect of treatment A (test) versus treatment B (control) on a time-to-event endpoint, the latter hereafter generically referred to as survival time. We let the denote survival time under treatment j so that

( ) = > is the true proportion of patients𝑇𝑇𝑗𝑗 in the overall target population with survival time𝑆𝑆𝑗𝑗 𝑡𝑡 greater𝑃𝑃𝑃𝑃� 𝑇𝑇than𝑗𝑗 𝑡𝑡t� for treatment j. The usual null hypothesis of interest is that the survival time distributions for the two treatments are the same, typically expressed as

: ( ) = ( ) for all t. (1)

0 𝐴𝐴 𝐵𝐵 With ( ) representing the true hazard𝐻𝐻 𝑆𝑆 𝑡𝑡function𝑆𝑆 𝑡𝑡 for treatment j, it is easy to see that

(ℎ𝑗𝑗) 𝑡𝑡 ( ) = ( )/ ( ) ( ). If ( ) and ( ) are proportional 𝑡𝑡 𝑡𝑡 ∫to0 eachℎ𝐴𝐴 𝑢𝑢 other𝑑𝑑𝑑𝑑�,∫ 0 (ℎ𝐵𝐵) 𝑢𝑢 𝑑𝑑 𝑑𝑑is the− time𝑙𝑙𝑙𝑙𝑙𝑙𝑆𝑆-𝐴𝐴invariant𝑡𝑡 −𝑙𝑙𝑙𝑙𝑙𝑙 hazard𝑆𝑆𝐵𝐵 𝑡𝑡 ratio≡ 𝜃𝜃 and𝑡𝑡 theℎ null𝐴𝐴 𝑡𝑡 hypothesisℎ𝐵𝐵 𝑡𝑡 can be written as 1-2 : = 1. The𝜃𝜃 logrank𝑡𝑡 ≡ 𝜃𝜃 test or, equivalently, the from the Cox proportional hazards 3 (PH)𝐻𝐻0 𝜃𝜃 model with only a treatment arm indicator, remains a popular option for testing H0 in randomized clinical trials.4-7 Two well-known issues emerge when the PH assumption is non- trivially violated, as often seen8-13 in practice: the power of the logrank test can be substantially diminished and the estimate from the Cox PH model can be hard to interpret.

One way to guard against a potential power loss associated with the logrank test under non-PH conditions is through the use of a weighted logrank test, commonly selected from the , class of

14 𝜌𝜌 ϒ tests proposed by Harrington and Fleming with weight function ( ) = { ( )} {1 𝐺𝐺 ( )} for 𝜌𝜌 𝛾𝛾 , 0, where ( ) is the Kaplan-Meier estimator of the pooled survival𝑤𝑤 𝑡𝑡 function𝑆𝑆̂ 𝑡𝑡 at− time𝑆𝑆̂ 𝑡𝑡 t. Note 0,0 that𝜌𝜌 𝛾𝛾 G≥ ≡ Z1 is 𝑆𝑆thê 𝑡𝑡 logrank which assigns equal weight to each event, while the G1,0 ≡ Z2, G1,1 ≡ Z3 and G0,1 ≡ Z4 place more weight on early, middle and late events, respectively. If prior knowledge about the expected nature of the between-treatment difference in the overall survival functions is available at the trial design stage, then a prudent choice between these four Z statistics, or selection of an alternate , statistic, can be made and pre-specified in the analysis 𝜌𝜌 ϒ plan. However, because such prior information𝐺𝐺 is usually not available, versatile combinations of weighted logrank tests into single overall tests have been proposed by several authors,15-21

2

including the MaxCombo test21 which uses the minimum of Z1, Z2, Z3 and Z4 as the test statistic when a one-tailed hypothesis test is planned and negative Zi’s indicate directional support for the test versus the control treatment.

Another way to mitigate the risk of reduced power associated with use of the logrank test under non-PH conditions, and to simultaneously avoid corresponding interpretational challenges with a single hazard ratio estimate, is to test based on an integrated weighted average of an estimate

22 of ( ) ( ) over a time interval (0,𝐻𝐻0τ]. A version of this test based on equal weights at each time𝑆𝑆𝐴𝐴 point,𝑡𝑡 − 𝑆𝑆 𝐵𝐵typically𝑡𝑡 referred to as the restricted survival time (RMST) test, has been popularized within the past decade by several authors.23-26 Note that ψ( ) = [ ( ) ( )] 𝜏𝜏 is the true between-treatment difference in mean survival time restricted𝜏𝜏 to the∫0 first𝑆𝑆𝐴𝐴 τ𝑡𝑡 time− 𝑆𝑆 𝐵𝐵units𝑡𝑡 𝑑𝑑of𝑑𝑑 follow-up after treatment initiation. In practice, ψ( ) is commonly estimated non-parametrically

as the difference between the area under the Kaplan𝜏𝜏-Meier curve up to time τ for each treatment. An advantage of the RMST approach is that the proportional hazards assumption is not required to deliver a clinically interpretable quantification of the comparative treatment effect. However, whether and by how much the RMST test improves upon the power of the logrank test depends on the choice of τ, on the patterns for the treatment arms being compared, and on the extent and nature of the departure from proportional hazards.

Combinations of weighted logrank tests and the RMST test are indeed rational alternatives to the logrank test when a departure from PH is anticipated. However, all three approaches share the following two shortcomings. First, they do not leverage a ubiquitous feature of randomized clinical trial populations, namely ‘structured’ patient heterogeneity (described below), and this can contribute to suboptimal power for testing . Second, even if the support rejection of ,

none of the aforementioned approaches are designed𝐻𝐻0 to readily deliver quantitative metrics that 𝐻𝐻aid0 in the assessment of whether the test treatment is likely to be survival-prolonging for all types of patients in the target population or only an identifiable subset.

3

The above observations motivate development of an alternate approach for in randomized clinical trials. To head in that direction, with = ( ), we begin by noting that

= ( ) is a statistically unambiguous causal estimand𝑌𝑌𝑗𝑗 ; 𝑙𝑙𝑙𝑙𝑙𝑙it represents𝑇𝑇𝑗𝑗 the expected within- patient∆ 𝐸𝐸 𝑌𝑌diff𝐴𝐴 −erence𝑌𝑌𝐵𝐵 in log survival time between treatments A and B if each patient could hypothetically be observed under each treatment. In a parallel arm randomized clinical trial, only one of and can be observed for each patient, but can still be estimated with minimal bias ( ) ( ) ( ) under standard𝑌𝑌𝐴𝐴 𝑌𝑌 𝐵𝐵assumptions since = ∆ . 𝐴𝐴 𝐵𝐵 𝐴𝐴 𝐵𝐵 𝐸𝐸 𝑌𝑌 − 𝑌𝑌 𝐸𝐸 𝑌𝑌 − 𝐸𝐸 𝑌𝑌

Next, we envision the overall target patient population as being a finite mixture of distinct subpopulations, which we refer to as ‘risk strata’. Higher to lower ordered risk strata comprise of patients with clinical prognoses of shorter to longer expected survival regardless of assigned treatment. Patients within a given risk stratum are prognostically homogeneous in that they have in common certain pre-treatment characteristics that jointly strongly associate with survival time. In statistical parlance, the survival times under a given treatment for patients within a risk stratum are presumed to follow a common distribution. With random variable denoting the true survival time under treatment j for risk stratum (1 ) and = ( 𝑇𝑇𝑖𝑖𝑖𝑖), we further envision that is distributed as + so that 𝑖𝑖 =≤ (𝑖𝑖 ≤ 𝑠𝑠 ) 𝑌𝑌represent𝑖𝑖𝑖𝑖 𝑙𝑙𝑙𝑙𝑙𝑙s𝑇𝑇 the𝑖𝑖𝑖𝑖 expected within-patient 27 𝑌𝑌difference𝑖𝑖𝑖𝑖 in log survival𝑌𝑌𝑖𝑖𝑖𝑖 time∆𝑖𝑖 under ∆treatments𝑖𝑖 𝐸𝐸 𝑌𝑌𝑖𝑖𝑖𝑖 A− 𝑌𝑌and𝑖𝑖𝑖𝑖 B within stratum i. This implies that ( ) = ( ), where ( ) = > is the true proportion of patients in risk stratum i

𝑆𝑆with𝑖𝑖𝑖𝑖 𝑡𝑡 survival𝑆𝑆𝑖𝑖𝑖𝑖 𝛾𝛾time𝑖𝑖𝑡𝑡 greater𝑆𝑆 than𝑖𝑖𝑖𝑖 𝑡𝑡 t under𝑃𝑃𝑃𝑃�𝑇𝑇 treatment𝑖𝑖𝑖𝑖 𝑡𝑡� j and = . We refer to as the true time ∆𝑖𝑖 ratio for patients in risk stratum i. It represents a𝛾𝛾 𝑖𝑖comparative𝑒𝑒 treatment𝛾𝛾𝑖𝑖 effect with a straightforward clinical interpretation; for example, = 1.25 that patients in risk stratum

i are expected to have 25% longer survival under treatment𝛾𝛾𝑖𝑖 A versus treatment B.

The conceptualization described above implies that the causal estimand can now be expressed as = , where is the proportion of patients in the entire target population that are in risk 𝑠𝑠 ∆stratum∑𝑖𝑖= i1 𝑓𝑓(𝑖𝑖∆𝑖𝑖 = 1𝑓𝑓)𝑖𝑖 . The null hypothesis of interest is reformulated as 𝑠𝑠 𝑖𝑖=1 𝑖𝑖 ∑ 𝑓𝑓 : = 0 (i.e., = 1) for all i (2) ∗ 𝐻𝐻0 ∆𝑖𝑖 𝛾𝛾𝑖𝑖 4

which is equivalent to

: [ ( ) = ( ) ] for all t. (3) ∗ 𝑠𝑠 0 𝑖𝑖=1 𝑖𝑖𝑖𝑖 𝑖𝑖𝑖𝑖 Note that if in (2) is true, then𝐻𝐻 ⋂ in 𝑆𝑆(1) is𝑡𝑡 also𝑆𝑆 true.𝑡𝑡 Furthermore, if all the ’s have the same ∗ sign (an assumption𝐻𝐻0 that we do not𝐻𝐻 require0 nor make), then is equivalent to ∆𝑖𝑖 ∗ 0 : = 0 (i. e. , = 𝐻𝐻1) , (4) ∗∗ 0 where = is the average (geometric𝐻𝐻 mean)∆ time ratio𝛾𝛾 for the overall population. ∆ 𝛾𝛾 𝑒𝑒

We propose the following approach to test the null hypothesis in (2) and to estimate the causal estimand (and subsequently ). A list of baseline covariates that have the potential to be prognostic∆ for survival under either𝛾𝛾 treatment is pre-specified in the analysis plan (Step 1). At the analysis stage, using all the observed survival times but blinded to patient-level treatment assignment, ‘noise’ covariates are removed with elastic net Cox regression (Step 2). The shortened covariate list is subsequently used by a conditional inference tree algorithm to segment the heterogeneous trial population into subpopulations of prognostically homogeneous patients (risk strata) (Step 3). After patient-level treatment unblinding, a treatment comparison is done within each formed risk stratum (Step 4) and stratum-level results are combined for overall statistical inference (Step 5).

Before proceeding further, it is important to differentiate the proposed strategy from the common use of the stratified logrank test for hypothesis testing and corresponding stratified Cox PH model for estimation based on pre-specified stratification factor(s). While this is generally a step in the right direction relative to the unstratified version of the logrank test and Cox PH model, we caution against this routine practice for two reasons. First, inclusion of a stratification factor that does not materially influence survival time under either treatment or exclusion of a stratification factor that does so can reduce power due to over-stratification or under-stratification, respectively, both due to model misspecification.28-29 Second, even with a proper choice of stratification factor(s), the stratified logrank test can suffer from a notable power loss either if the assumption of proportional hazards within each stratum is incorrect, or if the assumption is correct but the true hazard ratio is

5

not constant across the strata. The primary reason for the statistical inefficiency in the latter case is that the inverse- weighting scheme implicitly used to combine estimated stratum-level log hazard ratios for overall inference is suboptimal when there is a treatment by stratum on the hazard ratio scale.30 In our proposal, instead of using pre-stratification and a corresponding stratified logrank/Cox PH model analysis, we use unbiased post-stratification in tandem with a new analysis approach that is intended to boost power for detecting a true between- treatment difference in the distribution of survival times. Furthermore, for the primary analysis, we quantify comparative treatment effects using time ratios instead of hazard ratios, without any need for the proportional hazards assumption anywhere, and our null hypothesis and causal estimand of interest are clearly stated with a straightforward clinical interpretation.

The rest of this article is structured as follows. In Section 2, we provide additional details for our proposed 5-step stratified testing and amalgamation routine (5-STAR). We subsequently contrast its performance with that of the unstratified logrank/Cox PH model, [pre-]stratified logrank/Cox PH model and RMST comparison strategies using a hypothetical and two real clinical trial datasets in Section 3 and simulations in Section 4. Concluding remarks are provided in Section 5.

2. 5-STAR DETAILS

The main concept behind our proposed 5-STAR approach is depicted schematically in Figure 1. Additional details for each step are given below.

6

Figure 1. 5-STAR schema

Step 1: Pre-specify baseline covariates that may be prognostic for survival

This step entails pre-specifying in the statistical analysis plan, ideally in collaboration with relevant subject matter experts, a list of baseline covariates that have the potential to influence survival times under either treatment. The proposed methodology can easily accommodate a reasonably large number of candidate covariates of differing types (continuous, ordinal, or nominal), so more inclusivity is recommended. However, this should be done within bounds of sound statistical, clinical and operational judgement, such as avoiding the nomination of candidate covariates that are unlikely to be used in routine clinical practice due to high cost, patient inconvenience or other reasons. We assume all prognostic covariates that define the true risk strata described in the Introduction, while unknown, are included in the candidate set.

Step 2: Remove ‘noise’ covariates from the pre-specified candidate list

In this step, while still blinded to patient-level treatment assignment, covariates without sufficient evidence of an association with observed survival times are removed from the candidate list developed in Step 1. We considered two ways to implement this covariate filtering step: (i) random survival forest with variable importance measures and associated bootstrap confidence intervals31- 32 and (ii) elastic net Cox regression.33-34 Since (i) and (ii) had similar performance during the

7

initial stages of this research (results not shown), we chose the latter due to computational simplicity. The objective function for elastic net Cox regression is

… ; … , … | | + , (5) … 2 𝑝𝑝 1−𝜓𝜓 𝑝𝑝 2 1 𝑁𝑁 1 𝑝𝑝 1 𝑝𝑝 𝑘𝑘 𝑘𝑘 𝛽𝛽𝑚𝑚𝑚𝑚𝑚𝑚1 𝛽𝛽𝑝𝑝 �𝑁𝑁 𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙�𝑡𝑡 𝑡𝑡 𝑥𝑥 𝑥𝑥 𝛽𝛽 𝛽𝛽 � − 𝜆𝜆 �𝜓𝜓 ∑𝑘𝑘=1 𝛽𝛽 � 2 � ∑𝑘𝑘=1 𝛽𝛽 �� where ( ) is the Cox partial based on the pooled potentially censored survival

times (𝐿𝐿 ∙… ) and pre-specified baseline covariates ( … ), 0 is a tuning parameter and [0,𝑡𝑡11] is𝑡𝑡 𝑁𝑁a mixing parameter. In (5), = 0 and 𝑥𝑥1 =𝑥𝑥1𝑝𝑝 yield𝜆𝜆 ≥ objective functions for Cox 35 36 𝜓𝜓regression∈ with the well-known ridge and lasso𝜓𝜓 penalties,𝜓𝜓 respectively. The elastic net combines the strengths of the two approaches.

To identify the optimal and , we consider a grid of values {0.05, 0.1, 0.15, … , 0.95}. For each

fixed , we perform 10𝜓𝜓-fold cross𝜆𝜆 -validation to determine𝜓𝜓 the optimal value of . Here, optimal can be𝜓𝜓 defined either as the value of that directly minimizes the cross-validation𝜆𝜆 from the Cox partial likelihood model (denoted𝜆𝜆 as ) or as the largest value of which yields cross- validation error that is within one 𝜆𝜆𝑚𝑚𝑚𝑚 𝑚𝑚of the minimum deviance (denoted𝜆𝜆 as ); we use the former as a default for 5-STAR to reduce the risk of incorrectly eliminating prognostic𝜆𝜆1𝑠𝑠𝑠𝑠 covariates. Based on the optimized and , all covariates with a non-zero coefficient in the final

elastic net Cox regression model fit 𝜓𝜓are advanced𝜆𝜆 to Step 3.

Step 3: Segregate the overall population into subpopulations (risk strata) of prognostically homogeneous patients

After covariate filtering has been performed, still blinded to patient-level treatment assignment, the conditional inference tree (CTree) tool, an unbiased recursive partitioning algorithm developed by Hothorn et al,37 is used to segregate patients into well-separated risk strata. Briefly, for each covariate , a permutation test is performed to assess the null hypothesis that the distribution of

the pooled𝑋𝑋 𝑗𝑗logrank scores conditional on is the same as the marginal distribution of the pooled logrank scores, resulting in p-value . 𝑋𝑋If𝑗𝑗 at least one of the p-values is deemed statistically 38 significant after a Sidak multiplicity𝑝𝑝𝑗𝑗 adjustment, the algorithm splits the data based on the

8

covariate with the strongest association (i.e., with the smallest p-value). If the given covariate is binary, splitting into two nodes is straightforward. Otherwise, the split point is chosen to maximize the separation in the distribution of the logrank scores between the two forming nodes. The algorithm continues doing this separately under each formed node, switching between covariate selection and split determination. Newly formed nodes are grown in this manner until the null hypothesis of no association between the pooled logrank scores and any covariate in contention cannot be rejected or until predetermined criteria (e.g., requiring at least 40 patients per terminal node) are violated. Each terminal node represents a formed risk stratum.

The CTree algorithm is a very useful patient segmentation tool. However, given the nature of the covariate-based sample splitting, it is possible to see diverging nodes from secondary splits which have similar survival profiles, visualized as approximately overlapping Kaplan-Meier curves. This can result in potential over-stratification. To avoid this, the 5-STAR method considers two iterations of the CTree algorithm, named Step 3A and Step 3B. In Step 3A, all covariates surviving the elastic net filtering step are input into the CTree algorithm to form preliminary patient risk strata, as described above. These preliminary nodes are then sorted from highest to lowest risk based on the area under the overall Kaplan-Meier curve from time zero until the minimax survival time (i.e., the minimum of the maximum observed survival times over all treatment-pooled strata) for patients within a given preliminary risk stratum. This results in an ordinal stratum variable, with value 1 for patients in the highest risk preliminary risk stratum, value 2 for patients in the second highest risk preliminary risk stratum 2, and so on. At the start of Step 3B, all patients are considered to be part of a single final risk stratum and permutation testing is performed to determine if there is sufficient statistical evidence of an association between the ordinal stratum variable and the pooled logrank scores. As in Step 3A, this second run of CTree iteratively grows a tree, finding the best split point across the ordinal stratum variable until meeting the stopping criteria. Of note, a split can separate, for example, ordered preliminary strata (1, 2) from (3, 4) but not (1, 3) from (2, 4). If all ordered preliminary strata are deemed by the algorithm to have statistically different survival profiles, the final formed strata are the same as the preliminary strata (i.e., all possible splits occur in Step 3B). Otherwise, those that are not deemed statistically different are pooled in an order restricted manner to form a smaller number of final strata. The

9

multiplicity-adjusted p-value thresholds for allowing nodal splits need to be pre-specified; we use = 0.10 and = 0.20 as a default in Steps 3A and 3B, respectively.

3𝐴𝐴 3𝐵𝐵 𝛼𝛼 𝛼𝛼

At the end of Step 3A and 3B, still blinded to patient-level treatment assignment, suppose a total of final risk strata are formed. A graph with overlaid Kaplan-Meier curves for each of the c formed𝑐𝑐 risk strata can be helpful in judging the effectiveness of this critical risk-based patient segmentation step of 5-STAR; we show this with the three examples in the next Section.

Step 4: Treatment comparison within each formed risk stratum

In this step, patient-level treatment unblinding is done to enable a treatment comparison within each of the formed risk strata. Specifically, with denoting the log survival time for patient k

within formed risk stratum q (1 ), we consider𝑌𝑌𝑞𝑞𝑞𝑞 a basic log-linear form of the accelerated failure time model ≤ 𝑞𝑞 ≤ 𝑐𝑐 = + + (6)

𝑞𝑞𝑞𝑞 𝑞𝑞 𝑞𝑞 𝑞𝑞𝑞𝑞 𝑞𝑞 𝑞𝑞𝑞𝑞 where is a treatment indicator (1 for𝑌𝑌 treatment𝜇𝜇 𝛿𝛿 A 𝐼𝐼and 0𝜎𝜎 for𝜖𝜖 treatment B), is a random error

term with𝐼𝐼𝑞𝑞𝑞𝑞 an unknown density function, and and are intercept and𝜖𝜖𝑞𝑞𝑞𝑞 scale parameters, respectively. The true comparative treatment effect𝜇𝜇𝑞𝑞 for patients𝜎𝜎𝑞𝑞 in formed risk stratum q is . Note that each parameter is conditional on the overall population being represented as a mixture𝛿𝛿𝑞𝑞 of subpopulations𝛿𝛿𝑞𝑞 defined by the formed risk strata. While the formed risk strata will ultimately coincide with the true risk strata (conceptualized in the Introduction) with increasing sample size, this may not be the case for a given randomized clinical trial. Fortunately, this does not interfere with our main goal of estimation and inference for as long as we can obtain reliable estimates

for each ; we return to this point when we discuss ∆Step 5. 𝑞𝑞 𝛿𝛿

While non-parametric analysis options for (6) do exist,39 our preference, motivated by support from real datasets, is to use parametric approaches. Liao and Liu40 have reported that treatment- specific Kaplan-Meier curves formed from an overall (typically heterogenous) randomized clinical

10

trial patient population are often well approximated by parametric survival fits that assume survival times arise from a finite mixture of Weibull distributions. This mimics our own experience with the exploration of clinical trial datasets across different therapeutic areas. Given this, it is tempting to consider only a Weibull model fit for (6) in formed stratum q. However, we employ a model- averaging idea to add a layer of robustness against misspecification of the assumed underlying survival time distribution. Specifically, we recommend estimating through a Weibull model

(model 1), a log-normal model (model 2) and a log-logistic model (model𝛿𝛿𝑞𝑞 3), resulting in a point estimate , and corresponding variance , associated with model m=1,2,3. Here, the aforementioned𝛿𝛿̂𝑞𝑞 𝑚𝑚 Weibull, log-normal and log𝑉𝑉-𝑞𝑞logistic𝑚𝑚 distributions refer to the distribution of survival times, with corresponding distributions for log survival times, or more specifically, for the random error terms in (6) being the extreme value (Gumbel), normal and logistic, respectively. While other parametric survival time distributions (e.g., gamma) can also be considered for (6), our experience suggests that the aforementioned three will generally suffice. Either way, it is important to stress that the parametric distributions to be used in the model-averaging need to be pre-specified in the analysis plan.

The final point estimate of and corresponding variance are obtained using the following well-

41 known model-averaging formulas𝛿𝛿𝑞𝑞 based on M model fits:

. , = . (7) , −0 5𝐴𝐴𝐴𝐴𝐴𝐴𝑞𝑞 𝑚𝑚 , 𝑒𝑒 𝑀𝑀 −0 5𝐴𝐴𝐴𝐴𝐴𝐴𝑞𝑞 𝑚𝑚 𝑤𝑤𝑞𝑞 𝑚𝑚 ∑𝑚𝑚=1 𝑒𝑒 = , , (8) 𝑀𝑀 𝛿𝛿̂𝑞𝑞 ∑𝑚𝑚=1 𝑤𝑤𝑞𝑞 𝑚𝑚𝛿𝛿̂𝑞𝑞 𝑚𝑚

= , , + , 2. (9) 𝑀𝑀 2 𝑉𝑉𝑞𝑞 �∑𝑚𝑚=1 𝑤𝑤𝑞𝑞 𝑚𝑚�𝑉𝑉𝑞𝑞 𝑚𝑚 �𝛿𝛿̂𝑞𝑞 𝑚𝑚 − 𝛿𝛿̂𝑞𝑞� � Above, , is a measure of the goodness-of-fit for parametric model m, quantified using the

population𝐴𝐴𝐴𝐴𝐴𝐴 Akaike𝑞𝑞 𝑚𝑚 Information Criterion, and , is the associated weight assigned to the fit from model m, all within formed risk stratum q. Based𝑤𝑤𝑞𝑞 𝑚𝑚 on large-sample theory,

~ N(0,1) (10) �𝛿𝛿�𝑞𝑞−𝛿𝛿𝑞𝑞� �𝑉𝑉𝑞𝑞 11

so that an approximate 100 × (1 )% for is / . The point

estimate and confidence interval −for𝛼𝛼 the corresponding time ratio𝛿𝛿𝑞𝑞 (i.e.,𝛿𝛿̂𝑞𝑞 ∓ 𝑍𝑍𝛼𝛼=2�𝑉𝑉𝑞𝑞) are easily 𝛿𝛿𝑞𝑞 obtained using exponentiation. In addition to the confidence interval for𝛾𝛾 𝑞𝑞 , we𝑒𝑒 recommend reporting an estimate of the probability that treatment A prolongs expected 𝛾𝛾survival𝑞𝑞 relative to treatment B in formed risk stratum q, i.e., > 0 = > 1 , calculated using (10).

𝑞𝑞 𝑞𝑞 𝑃𝑃𝑃𝑃�𝛿𝛿 � 𝑃𝑃𝑃𝑃�𝛾𝛾 �

A natural alternative to the model in (6) is the popular Cox proportional hazards model. We prefer the former because there is no guarantee that the PH assumption will hold in each formed risk stratum, nor asymptotically in each of the true risk strata. There is, however, one exception which applies if the survival times for treatment B in formed risk stratum q follow a Weibull distribution and treatment A shifts the location of the log survival times under treatment B by , as in (6). In this special case, it can be shown that the following identities will hold: 𝛿𝛿𝑞𝑞 ( ) =

( ) + and ( ) = ( ) , where = / and = 𝑙𝑙𝑙𝑙𝑙𝑙is the�ℎ𝑞𝑞 familiar𝑞𝑞 𝑡𝑡 � 𝑞𝑞 𝜃𝜃 𝛽𝛽𝑞𝑞 𝑙𝑙𝑙𝑙𝑙𝑙hazard�ℎ𝑞𝑞 𝑞𝑞ratio.𝑡𝑡 � Given𝛽𝛽𝑞𝑞 our𝑆𝑆𝑞𝑞𝑞𝑞 observation𝑡𝑡 �𝑆𝑆𝑞𝑞𝑞𝑞 𝑡𝑡that� a Weibull𝛽𝛽𝑞𝑞 assumption−𝛿𝛿𝑞𝑞 𝜎𝜎𝑞𝑞 for 𝜃𝜃survival𝑞𝑞 𝑒𝑒 times within a prognostically homogeneous subpopulation will often (but not always) be reasonable, as a supplemental analysis, we recommend reporting a point estimate and confidence interval for each and an estimate of < 1 , all calculated using basic semi-parametric Cox PH model fits

𝜃𝜃within𝑞𝑞 each formed risk𝑃𝑃𝑃𝑃 stratum�𝜃𝜃𝑞𝑞 . �It should be recognized that these supplemental results will be hard to interpret if the PH assumption is clearly not supported by the data in one or more of the formed risk strata.

Step 5: Amalgamation of results across formed risk strata for overall hypothesis testing and estimation

After comparative treatment effect results within each formed risk stratum have been obtained, we are now ready to test in (2) and to estimate (and subsequently ). Let and N denote the ∗ number of patients in 𝐻𝐻formed0 risk stratum q and∆ the total number of𝛾𝛾 randomized𝑛𝑛𝑞𝑞 patients in the trial, respectively. The test statistics

12

= 𝑐𝑐 (11) ∑𝑞𝑞=1 𝑛𝑛𝑞𝑞𝛿𝛿�𝑞𝑞 𝐼𝐼 𝑐𝑐 2 𝑍𝑍 �∑𝑞𝑞=1 𝑛𝑛𝑞𝑞𝑉𝑉𝑞𝑞 and

= 𝑐𝑐 (12) ∑𝑞𝑞=1 𝑛𝑛𝑞𝑞𝑍𝑍𝑞𝑞 𝐼𝐼𝐼𝐼 𝑐𝑐 2 𝑍𝑍 �∑𝑞𝑞=1 𝑛𝑛𝑞𝑞 are natural choices for testing , where = / . Asymptotically, both and are ∗ distributed as N(0,1) under . 𝐻𝐻Intuitively,0 𝑍𝑍for𝑞𝑞 a one𝛿𝛿̂𝑞𝑞 -�tailed𝑉𝑉𝑞𝑞 test in the direction favoring𝑍𝑍𝐼𝐼 the𝑍𝑍𝐼𝐼𝐼𝐼 test ∗ treatment, (11) will generally𝐻𝐻 0be more powerful than (12) if larger are expected a priori to be associated with larger , such as when lower risk patients, who may𝛿𝛿̂ 𝑞𝑞have fewer events and at an overall slower rate, experience𝑉𝑉𝑞𝑞 greater relative benefit from the test treatment. Since this type of information is typically unavailable at the design stage of the trial, instead of adopting either or

as a default, we recommend using 𝑍𝑍𝐼𝐼 𝐼𝐼𝐼𝐼 𝑍𝑍 = ( , ) (13)

𝑚𝑚𝑚𝑚𝑚𝑚 𝐼𝐼 𝐼𝐼𝐼𝐼 as the test statistic. The exact42 probability𝑍𝑍 density𝑚𝑚𝑚𝑚𝑚𝑚 function𝑍𝑍 𝑍𝑍 (PDF) of is

𝑚𝑚𝑚𝑚𝑚𝑚 ( ) = 2 ( ) , 𝑍𝑍 (14) 1−𝜌𝜌 2 𝑓𝑓 𝑧𝑧𝑚𝑚𝑚𝑚𝑚𝑚 𝜙𝜙 𝑧𝑧𝑚𝑚𝑚𝑚𝑚𝑚 Φ ��1−𝜌𝜌 𝑧𝑧𝑚𝑚𝑚𝑚𝑚𝑚� where is the true correlation between and ; (. ) and (. ) are the density function and

cumulative𝜌𝜌 distribution function of the standard𝑍𝑍𝐼𝐼 normal𝑍𝑍𝐼𝐼𝐼𝐼 𝜙𝜙 distributionΦ , respectively. In practice, is typically very close to one and can be estimated remarkably well with 𝜌𝜌

= 𝑐𝑐 2 . (15) ∑𝑞𝑞=1 𝑛𝑛𝑞𝑞�𝑉𝑉𝑞𝑞 𝑐𝑐 2 𝑐𝑐 2 𝜌𝜌� �∑𝑞𝑞=1 𝑛𝑛𝑞𝑞𝑉𝑉𝑞𝑞�∑𝑞𝑞=1 𝑛𝑛𝑞𝑞 An asymptotic one-tailed p-value (p) for testing in (2) is easily calculated using (11) to (15) ∗ and the null hypothesis is rejected if p < α/2 (= 0.025𝐻𝐻0 by default). In large samples, rejection of ∗ can be interpreted as reliable statistical evidence of treatment A prolonging survival relative𝐻𝐻 to0 treatment B, on average, for patients in at least one of the formed risk strata, and hence by extension in at least one of the true risk strata. The justification for the latter extension is based on the following two lines of reasoning. First, because the formed strata and true strata represent almost

13

the same (if not identical) risk-based groupings of patients asymptotically. Second, a stratified analysis based on formed risk strata and a corresponding analysis based on (hypothetically known) true risk strata implicitly address the same estimand, , which is zero under . This follows ∗ because, in a typical analysis context, adjustment∆ for a different𝐻𝐻 0set of baseline covariates changes the estimate but not the estimand.43

The 5-STAR point estimate of is obtained as

∆ = 𝑐𝑐 (16) ∑𝑞𝑞=1 𝑤𝑤𝑞𝑞𝛿𝛿�𝑞𝑞 𝑐𝑐 ∆� ∑𝑞𝑞=1 𝑤𝑤𝑞𝑞 with asymptotic variance

= 𝑐𝑐 2 (17) ∑𝑞𝑞=1 𝑤𝑤𝑞𝑞V𝑞𝑞 2 𝑐𝑐 𝑉𝑉�∆�� ∑𝑞𝑞=1 𝑞𝑞 where = if = and = /� 𝑤𝑤if � = . An asymptotic 100 × (1 )%

𝑤𝑤𝑞𝑞 𝑛𝑛𝑞𝑞 𝑧𝑧𝑚𝑚𝑚𝑚𝑚𝑚 𝑧𝑧𝐼𝐼 𝑤𝑤𝑞𝑞 𝑛𝑛𝑞𝑞 �𝑉𝑉𝑞𝑞 𝑧𝑧𝑚𝑚𝑚𝑚𝑚𝑚 𝑧𝑧𝐼𝐼𝐼𝐼 − 𝛼𝛼 confidence interval for is calculated as , / where , / is the relevant

𝑚𝑚𝑚𝑚𝑚𝑚 𝛼𝛼 2 𝑚𝑚𝑚𝑚𝑚𝑚 𝛼𝛼 2 quantile based on the distribution∆ in (14). Finally∆� ∓ ,𝑍𝑍 point and� 𝑉𝑉confidence�∆�� interval𝑍𝑍 estimates for are

obtained by exponentiating their counterparts for described above. Corresponding results𝛾𝛾 for = , where = , can be easily obtained∆ for a supplemental analysis, as needed, using 𝛽𝛽 𝑠𝑠 𝜃𝜃basic𝑒𝑒 Cox PH model𝛽𝛽 ∑ fits𝑖𝑖=1 with𝑓𝑓𝑖𝑖𝛽𝛽𝑖𝑖 each of the formed risk strata and analogs of the formulas described above.

3. THREE ILLUSTRATIVE EXAMPLES To help readers check their understanding of the 5-STAR method, and to illustrate its utility, we walk through its application in great detail using a hypothetical dataset, followed by application to two real datasets. The three examples differ primarily in terms of sample size, cardinality of the candidate set of baseline covariates in Step 1 of 5-STAR, and/or evidence of non-proportional hazards in the overall (i.e., unstratified) trial population.

14

Example 1 (hypothetical clinical trial)

Consider a hypothetical clinical trial in which 600 patients were randomized with equal probability to either a test treatment (A) or a control treatment (B), without pre-stratification on any factor. Assuming proportional hazards, suppose that a clinically meaningful hazard ratio of 0.80 or less, loosely interpreted by some as ‘at least a 20% risk reduction’, was hoped for. Based on a simulated dataset, Figure 2 (left panel) shows Kaplan-Meier curves for each treatment along with a logrank test one-tailed p-value of 0.105 and estimated hazard ratio (95% CI) of 0.87 (0.70, 1.08) obtained from a basic Cox PH model. These results look both statistically and clinically disappointing. However, a crossing of the smoothed estimates of the log hazard functions and a p-value of 0.015 from the popular Grambsch and Therneau [GT] test44 (right panel) provide cautionary evidence of non-proportional hazards. Unfortunately, even if non-PH was anticipated a priori in this hypothetical setting and one of the two alternatives to the logrank test discussed in the Introduction had been pre-specified in the analysis plan, the result would still have been disappointing: one- tailed p-values of 0.057 and 0.182 for the MaxCombo and RMST test, respectively. In essence, application of the common approaches in current practice would have resulted in a ‘negative’ trial due to lack of statistical evidence of an overall survival benefit of the test versus the control treatment. We now show how the application of 5-STAR here leads to a different and more appropriate conclusion.

15

Figure 2. Kaplan-Meier curves for each treatment (left) and corresponding smoothed versions of estimated log hazard functions (right) for Example 1.

Suppose 50 baseline covariates, generically labeled X1-X50, were pre-specified in Step 1 and included in the hypothetical dataset. Of these, based on elastic net Cox regression applied to the

pooled survival times, i.e., blinded to patient-level treatment assignment, 6 binary covariates (X1,

X2, X6, X7, X8, X15) and 4 continuous covariates (X26, X31, X38, and X40) are deemed potentially prognostic for survival in Step 2. For completeness, salient details of the optimization of the elastic net mixing and tuning parameters, ( , ), and resulting ‘solution path’ are shown graphically in

the left and right panels of Figure 3, 𝜓𝜓respectively𝜆𝜆 .

16

Figure 3. Graphical details for the elastic net Cox regression filtering step (Example 1). Left: each curve represents a mixing parameter and each dot represents the tuning parameter value with the smallest deviance. Here ( , ) = (0.95, 0.04) is chosen to minimize deviance. Right: solution path for the chosen value, showing how the coefficients𝜓𝜓 for the elastic net model fit change over the 𝜆𝜆 of . Of the 50 input covariates,𝜓𝜓 ten𝜆𝜆 (X1, X2, X6, X7, X8, X15, X26, X31, X38 and X40) have a non-zero coefficient at the 𝛼𝛼optimal value (shown via dotted vertical line) 𝜆𝜆

𝜆𝜆

Based on the CTree algorithm results, Figure 4 reveals the formation of six preliminary (top panel; Step 3A) and four final (bottom panel; Step 3B) risk strata defined by three baseline

covariates: X1 (binary), X2 (binary) and X26 (continuous, with a 0.35 cut-point). The overall Kaplan-Meier curves pooled across treatment arms for the four final risk strata are clearly well- separated, with survival times steadily increasing when transitioning in order from the highest risk stratum (S1) to the lowest risk stratum (S4).

Having reached Step 4 in the 5-STAR method, patient-level treatment unblinding can now be done. The resulting Kaplan-Meier curves for both treatments are shown within each formed risk stratum in the top panel of Figure 5. The bottom panel displays point estimates and 95% CIs for stratum- level time ratios, along with corresponding estimates of the probability that the time ratio is greater than one. All these quantities were calculated using expressions (6) through (10).

17

Figure 4. Risk strata formation for Example 1 using the conditional inference tree algorithm blinded to patient-level treatment assignment. Left: results from Step 3A (preliminary risk strata) and Step 3B (final risk strata). Right: corresponding Kaplan-Meier curves for each risk stratum.

18

Figure 5. Results from 5-STAR Step 4 and Step 5 for Example 1. Top: Kaplan-Meier curves by treatment within each formed risk stratum. Bottom: showing stratum-level results for each formed risk stratum as well as the overall (i.e., stratum-averaged) result. TR = time ratio, with TR > 1 favoring the test treatment. HR = hazard ratio, with HR < 1 favoring the test treatment.

19

Finally, in Step 5 of 5-STAR, the individual formed risk stratum results are combined using expressions (11) through (17) leading to the estimated average time ratio (95% CI) of 1.14 (1.05, 1.24) shown in the bottom panel of Figure 5. Based on = 3.05, = 2.95 and = 0.992, the one-tailed p-value for 5-STAR is 0.001, providing strong𝑍𝑍𝐼𝐼 statistical𝑍𝑍 evidence𝐼𝐼𝐼𝐼 that the𝜌𝜌� test treatment prolongs survival relative to the control treatment, on average. This conclusion is different from that reached by the logrank, MaxCombo and RMST comparison tests.

Importantly, while 5-STAR has detected an efficacy signal favoring the test treatment over the control treatment in an ‘overall’ sense, it does not automatically imply that the test treatment should be deemed a better efficacy option for all types of patients. We feel it is necessary to look for subgroup(s) of patients defined by the formed risk strata that may have a ‘concerningly’ small likelihood of longer survival for the test treatment versus the control treatment; among several subjective options, we recommend using ( > 1) less than 20% as a flagging mechanism.

Based on the results shown in Figure 5, since𝑃𝑃𝑃𝑃 none𝑇𝑇𝑇𝑇 of the formed risk strata get flagged, it seems reasonable to conclude that the test treatment is a generally better option than the control treatment for longer survival, on average, for patients across all risk strata in the target population.

As a supplemental analysis, within each formed risk stratum, a point estimate and confidence interval for the hazard ratio along with a corresponding estimate of ( < 1) are shown in the bottom panel of Figure 5. Also reported is the estimated average HR 𝑃𝑃𝑃𝑃(95%𝐻𝐻𝐻𝐻 CI) of 0.72 (0.57, 0.92); note that the upper bound of this CI is less than one and the point estimate is better than the aforementioned clinically meaningful threshold of 0.80. Importantly, these additional results are interpretable because no obvious departure from PH is detected within any formed risk stratum (GT p-values: 0.127, 0.987, 0.537, 0.507). Furthermore, it is reassuring to see that conclusions based on time ratios and hazard ratios are reasonably well aligned.

Example 2 (real clinical trial; oncology therapeutic area)

20

We now consider a real Phase III randomized clinical trial described in Lipkovich et al.45 A total of 599 patients with a hematological malignancy were enrolled, with 303 receiving the experimental treatment and the remaining 296 receiving the control treatment. As no stratification factors were pre-specified for or analysis, the [unstratified] logrank test was used for the primary analysis. This analysis missed the one-sided significance threshold of 0.025 with an overall one-tailed p-value of 0.035. The corresponding hazard ratio (95% CI), as estimated via an unstratified Cox PH model, was 0.85 (0.71, 1.01). Upon examining the Kaplan-Meier curves and estimated smoothed log hazards for each treatment (left and middle panels of Figure 6, respectively), we see hints of non-proportional hazards, with a separation in the curves occurring around the middle of the time points and crossing log hazard functions. The GT test yields a p- value of 0.002, providing statistical evidence against proportional hazards. Unlike the hypothetical example, here the MaxCombo and RMST approaches do generate a retrospective statistical ‘win’ for the trial with one-tailed p-values of 0.004 and 0.014, respectively. However, they are not designed to deliver a reliable and interpretable estimate of the comparative treatment , either overall or in relevant subgroups of patients.

We now illustrate the application of 5-STAR to these data. In the study, 14 baseline covariates were recorded, including both nominal covariates (patient sex, race, prior therapy outcome and presence/absence of nine cytogenetic markers) and ordinal covariates (cytogenetic category and IPSS-R prognostic risk score). All these are included in Step 1. In Step 2, seven cytogenetic markers get filtered out using the elastic net Cox regression model; the remaining seven covariates are passed into the CTree algorithm, showing evidence of a possible prognostic association with survival. In Step 3A, five preliminary strata are formed. No pooling is done in Step 3B, leaving the same five final risk strata. The formed risk strata are defined using the following covariates: cytogenetic marker 6 (Cytogen6, Present (1) vs. Absent (0)), outcome of prior therapy (Priorout, with split between higher risk category “Progress” and lower risk categories {“Failure”, “Relapse”}), IPSS-R prognostic score (IPSS, with split between higher risk score groups {High (3), Very High (4)} and lower risk score groups {Low (1), Intermediate (2)}), and cytogenetic category (Cytogencat, with split between lower risk group {Very Good (1), Good (2) , Intermediate (3)} and higher risk group {Poor (4), Very Poor (5)}). The Kaplan-Meier curves for the five final

21

risk strata are shown in Figure 6 (right panel). While visually there may appear to be some overlap within two pairs of curves (medium risk strata S2-S3 and low risk strata S4-S5), there is a clear separation between the three groups of curves, a corresponding clear increase in median survival, and strong separation at early time points between the aforementioned curve pairs.

After the final risk strata are formed, the within-stratum treatment effects are estimated in Step 4. The treatment-specific Kaplan-Meier curves for each formed stratum (top panel) and corresponding forest plot of estimated stratum-level time ratios and 95% confidence intervals (bottom panel) are provided in Figure 7. At the end of Step 5, the estimated average time ratio (95% CI) is 1.23 (1.08, 1.40). Furthermore, based on = 3.15, = 2.74 and = 0.990, the

corresponding one-tailed p-value is 0.001. This shows𝑍𝑍 𝐼𝐼strong evidence𝑍𝑍𝐼𝐼𝐼𝐼 that the test𝜌𝜌� treatment is beneficial in extending survival time, on average, i.e., for at least a subgroup of patients, relative to the control treatment. As in the previous example, we now take a closer look at the formed risk stratum-level results. Here, for the higher risk patients (S1-S3, corresponding to about 50% of the patients) the test treatment is estimated to prolong expected survival by an impressive 50-85% over the control treatment, with greater than 99% probability that the true time ratio exceeds 1. In contrast, there is no evidence that the test treatment will be more beneficial for the lowest risk patients (S5) where ( > 1) is < 5%. In a regulatory setting, it would seem reasonable to question the approvability𝑃𝑃𝑃𝑃 𝑇𝑇𝑇𝑇 of the test treatment for this 12% subset of the target patient population, corresponding to patients with lower IPSS risk scores and better cytogenetic categories.

22

Figure 6. Kaplan-Meier curves for each treatment (left) and corresponding smoothed versions of estimated log hazard functions (middle) for Example 2. Right: KM curves for final risk strata formed using the conditional inference tree algorithm blinded to patient-level treatment assignment.

23

Figure 7. Results from 5-STAR Step 4 and Step 5 for Example 2. Top: Kaplan-Meier curves by treatment within each formed risk stratum. Bottom: forest plot showing stratum-level results for each formed risk stratum as well as the overall (i.e., stratum-averaged) result. TR = time ratio, with TR > 1 favoring the test treatment. HR = hazard ratio, with HR < 1 favoring the test treatment.

24

For the most part, conclusions are consistent when examining the supplemental hazard ratio results. The overall estimated average HR (95% CI) is 0.81 (0.66, 0.99), which is more promising both statistically and clinically compared to the corresponding result from the prespecified analysis (Cox PH model). Of note is the second highest formed risk stratum S2, where the estimated hazard ratio (95% CI) is 0.94 (0.69, 1.30) with less than 62% probability that the true hazard ratio is less than one. This shows materially lower evidence of test treatment versus control treatment benefit as compared to that based on the time ratio-based estimate. However, this stratum, along with strata S1 and S3 show evidence of non-PH (GT p-value < 0.02 in all three strata, and < 0.0001 in S2); this makes the hazard ratio results hard to interpret, amplifying the utility of the corresponding primary analysis results using time ratios.

Example 3 (real clinical trial; cardiovascular therapeutic area)

As a final example, we consider a large phase III cardiovascular clinical trial.46 This randomized study enrolled 18,144 patients who had recently experienced an acute coronary syndrome: 9,067 received the test treatment and 9,077 received the control treatment (standard of care). Randomization was stratified based on three pre-defined baseline factors: PLL (prior use of lipid lowering therapy, a binary variable indicating whether or not a patient had used statins before trial initiation), ACSD (type of acute coronary syndrome experienced, STEMI (ST-Elevation Myocardial Infarction) or non-STEMI), and EACS (representing patient participation status in a previous cardiovascular trial: unenrolled, enrolled receiving test treatment, or enrolled receiving control treatment). As only non-STEMI patients were enrolled in the EACS trial, eight pre-defined strata were used for randomization and subsequent protocol-defined analyses.

For illustrative purposes, we focus on the time to stroke (fatal or non-fatal), a clinically important exploratory endpoint for the trial with over 95% censoring. The Kaplan-Meier curves for each treatment arm are shown in the left panel of Figure 8; unlike the previous two examples, here there is no evidence of non-proportional hazards, with a GT p-value of 0.479. In the middle panel, we provide the overall (i.e., pooled across treatment arms) Kaplan-Meier curves for each of the eight pre-defined strata. There is a substantial amount of overlap in some of the curves, indicating a

25

potentially suboptimal choice of pre-stratification factor(s) leading to over-stratification. The pre- specified stratified logrank test just falls short of at the one-sided 0.025 alpha level, with a p-value of 0.026 and corresponding HR (95% CI) of 0.86 (0.73, 1.00) as estimated via the stratified Cox PH model. The corresponding one-tailed p-values for the RMST comparison and MaxCombo test are 0.024 and 0.039, respectively.

We now apply the 5-STAR method to this large dataset. Forty-five potentially prognostic variables are specified in Step 1, including demographic, clinical disease history, and baseline lipid level information. Of these, blinded to patient-level treatment assignment, 22 pass the filtering step in Step 2 as having some evidence of association with survival time. Finally, in Step 3, five preliminary and four final risk strata are formed based on continuous variable age (with cut-point at 67 years) and three binary patient history variables: HSCD (history of cerebrovascular disease), HSAF (history of atrial fibrillation), and PRMI (prior myocardial infarction). Overall Kaplan- Meier curves for the four final formed risk strata are shown in the right panel of Figure 8. There is a much more distinct separation in the pooled survival curves compared to that in the plot with the curves based on the pre-specified strata (middle panel). In particular, the highest formed risk stratum has clearly worse prognosis compared to the other three, which still have close to no overlap. This example vividly illustrates how the objective risk-based patient segmentation component of 5-STAR can reveal ‘structured’ patient heterogeneity using an algorithm that is blinded to patient-level treatment assignment. This type of information, important both from a clinical and statistical perspective, often remains hidden when viewed through strata defined by (often subjectively chosen) pre-stratification factors at the design stage of the trial.

Kaplan-Meier curves for both treatments within each formed risk stratum (top panel) and a corresponding forest plot of the estimated comparative treatment effects (bottom panel) are shown in Figure 9, illustrating the components behind the overall comparative treatment effect estimate obtained in Step 5 of 5-STAR. The average estimated time ratio (95% CI) is 1.30 (1.04, 1.81), indicating that, on average, patients on the test treatment have 30% longer stroke-free time compared to those on the control treatment. Based on = 2.32, = 2.36 and = 0.998, the

corresponding one-tailed p-value is 0.010, indicating statistical𝑍𝑍𝐼𝐼 evidence𝑍𝑍𝐼𝐼𝐼𝐼 of efficacy𝜌𝜌� benefit from 26

the test treatment compared to the control for at least some patients. Looking at the results within the formed risk strata, the lowest risk patients (i.e., those that are less than 67 years of age, with no prior MI and no history of cerebrovascular disease [S4]), representing just over half of all patients, have the largest apparent gain from taking the test treatment with an estimated time ratio (95% CI) of 1.50 (1.04, 1.61) and 99% probability of improved survival benefit (Pr( > 1)) relative to the control treatment. The supplemental hazard ratio-based results are 𝑇𝑇𝑇𝑇clinically consistent with the time ratio results, with average hazard ratio (95% CI) of 0.81 (0.67, 0.97), and an estimated hazard ratio (95% CI) in formed risk stratum S4 of 0.70 (0.55, 0.90) with over 99% probability that the true hazard ratio is less than one for such patients. Of note, none of the Pr( > 1) or Pr( < 1) values shown in Figure 9 are even close to being lower than 20%, our subjective𝑇𝑇𝑇𝑇 threshold𝐻𝐻 𝐻𝐻of concern introduced in Example 1. Accordingly, if these results were to serve as the primary basis for a regulatory action, we believe it would be reasonable to consider approvability of the test treatment for patients across all risk strata.

27

Figure 8. Kaplan-Meier (KM) curves for Example 2. Left: KM curves for test and control treatments. Middle: KM curves for each pre-specified stratum defined in the study protocol. Right: KM curves for final risk strata formed using the conditional inference tree algorithm blinded to patient-level treatment assignment.

28

Figure 9. Results from 5-STAR Step 4 and Step 5 for Example 3. Top: Kaplan-Meier curves by treatment within each formed risk stratum. Bottom: forest plot showing stratum-level results for each formed risk stratum as well as the overall (i.e., stratum-averaged) result. TR = time ratio, with TR > 1 favoring the test treatment. HR = hazard ratio, with HR < 1 favoring the test treatment.

29

4. SIMULATION STUDY 4.1 Set-up We simulated a clinical trial setting in which 600 patients are randomized in a 1:1 ratio to receive either a test treatment (A) or control treatment (B). For each patient, 50 correlated baseline covariates, - binary and - continuous, are measured. The heterogeneous patient population consist𝑋𝑋1 𝑋𝑋25s of four distinct𝑋𝑋26 𝑋𝑋prognostic50 subpopulations (risk strata) defined by a joint composite of two binary covariates ( and ) and one dichotomized continuous covariate (

0.4), as shown in Table 1. We set the𝑋𝑋 1true marginal𝑋𝑋2 means of the binary and continuous covariates𝑋𝑋26 ≤ to be 0.5 and 0, respectively. Furthermore, we assumed a true pairwise correlation of 0.2 between the three aforementioned prognostic covariates and sampled from (0, = 0.15) for all other pairwise correlations; these correlations were motivated by real𝑁𝑁 datasets𝑠𝑠𝑠𝑠 across different therapeutic areas (details omitted).

Table 1 Simulation conditions showing four true risk strata defined by three baseline covariates, and median survival time for the control treatment, hazard ratio (HR) and time ratio (HR) within each true risk stratum

Simulation Scenarios and Stratum-level Median Treatment Effects Risk Survival X1 X2 X26 Null Alt-1 Alt-2 Alt-3 Stratum (control HR = 1 Equal HRs Increasing HRs Decreasing HRs treatment) HR TR HR TR HR TR HR TR 0 0 ≤ 0.4 6.0 S1 1 1 0.7 1.15 0.42 1.42 0.95 1.02 0 1 ≤ 0.4 months 0 0 > 0.4 8.4 S2 1 1 0.7 1.13 0.7 1.13 0.86 1.05 1 0 ≤ 0.4 months 0 1 > 0.4 10.8 S3 1 1 0.7 1.11 0.86 1.04 0.7 1.11 1 1 ≤ 0.4 months 1 0 > 0.4 13.2 S4 1 1 0.7 1.09 0.95 1.01 0.42 1.24 1 1 > 0.4 months

For each treatment group, the random number of patients enrolled from each of the four risk strata was sampled from a ( = 300, = = = = 0.25) distribution. (Of note,

simulations with unequal𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚 prevalence𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚 𝑛𝑛 across the𝑝𝑝 risk1 strata𝑝𝑝2 𝑝𝑝wer3 e also𝑝𝑝4 conducted and yielded similar conclusions; details omitted). A trial entry time for each patient = 1, … , within risk stratum

𝑘𝑘 𝑛𝑛𝑖𝑖 30

= 1, … ,4, ordered from highest risk to lowest risk, and treatment = , was generated as

𝑖𝑖 ~ (0, ) where = 0.75 years, or 9 months. True survival𝑗𝑗 𝐴𝐴 times𝐵𝐵 were generated from𝑒𝑒𝑖𝑖𝑖𝑖𝑖𝑖 𝑈𝑈a𝑈𝑈𝑈𝑈𝑈𝑈𝑈𝑈𝑈𝑈𝑈𝑈 Weibull distribution𝐸𝐸 𝐸𝐸 with shape and scale parameters determined by risk stratum membership and treatment assignment as Weibull = , = , where =

( , , , ) = (2.5, 3, 3.5, 4), and 𝑆𝑆back𝑖𝑖𝑖𝑖𝑖𝑖 ∼-calculated� 𝑠𝑠fromℎ𝑎𝑎𝑎𝑎𝑎𝑎 the𝜅𝜅 shape𝑖𝑖 𝑠𝑠𝑠𝑠𝑠𝑠𝑠𝑠𝑠𝑠 and 𝜂𝜂desired𝑖𝑖𝑖𝑖� median𝜿𝜿 survival𝜅𝜅1 𝜅𝜅2 𝜅𝜅 3 time𝜅𝜅4 for the control treatment𝜂𝜂𝑖𝑖𝑖𝑖 arm (in years) of each stratum, = 𝐵𝐵 ( , , , ) = (0.5, 0.7, 0.9, 1.1), as = × ln(2) 1 . Scale parameters in the𝒎𝒎 test −𝜅𝜅𝑖𝑖 𝑚𝑚1𝐵𝐵 𝑚𝑚2𝐵𝐵 𝑚𝑚3𝐵𝐵 𝑚𝑚4𝐵𝐵 𝜂𝜂𝑖𝑖𝑖𝑖 𝑚𝑚𝑖𝑖𝑖𝑖 treatment arm were calculated as = × 1 , where was the hazard ratio for stratum . −𝜅𝜅𝑖𝑖 This set-up ensured proportional hazards𝜂𝜂𝑖𝑖𝑖𝑖 𝜂𝜂 in𝑖𝑖𝑖𝑖 truth𝜃𝜃𝑖𝑖 within each𝜃𝜃 𝑖𝑖risk stratum. 𝑖𝑖

In each simulated trial, patients were followed until 330 deaths had accrued. This target event total is often chosen in practice because it gives approximately 90% power for the logrank test when the proportional hazards assumption holds, the true hazard ratio is 0.7, and testing is done at the one-tailed alpha=0.025 level. The ‘observed’ simulated data for each trial were ( , , ),

where = min ( , ) was the observed survival time, was the random𝑇𝑇𝑖𝑖 𝑖𝑖𝑖𝑖time𝛿𝛿𝑖𝑖 𝑖𝑖𝑖𝑖at which𝑥𝑥𝑖𝑖𝑖𝑖𝑖𝑖 330 events𝑇𝑇𝑖𝑖𝑖𝑖𝑖𝑖 had accrued,𝑆𝑆𝑖𝑖𝑖𝑖𝑖𝑖 𝐶𝐶 indicating− 𝑒𝑒𝑖𝑖𝑖𝑖𝑖𝑖 end of patient follow-up, 𝐶𝐶= > was the censoring indicator, equal to 0 for all patients whose true event𝛿𝛿𝑖𝑖 𝑖𝑖𝑖𝑖time 𝐼𝐼wa�𝑆𝑆s𝑖𝑖𝑖𝑖𝑖𝑖 observed𝐶𝐶 − 𝑒𝑒 and𝑖𝑖𝑖𝑖𝑖𝑖� 1 for all patients who were censored, and was the vector of patient-level baseline covariate data.

𝑖𝑖𝑖𝑖𝑖𝑖 𝑥𝑥

Four scenarios were considered to evaluate the performance of the competing methods under different patterns of hazard ratios across the risk strata. First, to evaluate type I error, a null scenario was considered in which there was no treatment difference in any of the risk strata, i.e., = ( ) ( ) , , , = 1,1,1,1 . Three alternative scenarios were also considered, all with an average𝜽𝜽 log𝜃𝜃1 hazard𝜃𝜃2 𝜃𝜃3 𝜃𝜃ratio4 of = = (0.7), with = , as noted in Section 2. In Alt-1 we set 𝑠𝑠 𝛽𝛽𝑖𝑖 = (0.7, 0.7, 0.7, 0𝛽𝛽.7), ∑i.e.𝑖𝑖=,1 equal𝑓𝑓𝑖𝑖𝛽𝛽𝑖𝑖 hazard𝑙𝑙𝑙𝑙 ratios across𝜃𝜃𝑖𝑖 the𝑒𝑒 ordered risk strata. In Alt-2 we set = 𝜽𝜽(0.42, 0.7, 0.86, 0.95), i.e., increasing hazard ratios, implying larger survival benefit of the 𝜽𝜽test versus the control treatment for patient subpopulations with higher risk. Finally, in Alt-3 we set = (0.95, 0.86, 0.7, 0.42), i.e., decreasing hazard ratios, implying larger survival benefit of the 31 𝜽𝜽 test versus the control treatment for patient subpopulations with lower risk. All the scenarios are summarized in Table 1. In addition to the true hazard ratios for each risk stratum, we have also shown the corresponding true time ratios calculated as = , where = / . ∆𝑖𝑖 𝑖𝑖 𝑖𝑖 𝑖𝑖 𝑖𝑖 𝛾𝛾 𝑒𝑒 ∆ −𝛽𝛽 𝜅𝜅

We compared the performance of 5-STAR (both time ratio and hazard ratio versions) with that of the unstratified logrank test/Cox PH model, a misspecified stratified logrank test/Cox PH model, the MaxCombo and the RMST comparison methods. The misspecified stratified logrank test/Cox PH model was intended to mimic a realistic setting in which expert knowledge may guide researchers to correctly identify only a subset of the true prognostic covariates at the trial planning stage; in effect, either some prognostic covariates are missed, suboptimal cut-offs are selected for dichotomization of prognostic continuous covariates, or non-prognostic covariates are erroneously used for pre-stratification. Here, our pre-specified stratified Cox model analysis reflected strata defined by ‘correct’ covariates and (the latter discretized using a slightly suboptimal cut-

off of zero instead of the truly optimal𝑋𝑋2 0.4)𝑋𝑋26 as well as an incorrect, i.e., non-prognostic, covariate .

3 𝑋𝑋

In each of the four simulation scenarios, for each competing method, we computed the proportion of times in 20,000 simulated trials that the null hypothesis in (2), equivalent in this setting to ∗ in (4) and to in (1), was rejected at the one-sided 2.5%𝐻𝐻0 significance level incorrectly (for ∗∗ type𝐻𝐻0 I error) or correctly𝐻𝐻0 (for power). For 5-STAR, we also calculated mean percent bias for the average time ratio ( ) and average hazard ratio ( ) estimands along with the corresponding coverage of the 95% 𝛾𝛾CI for each parameter; these metrics𝜃𝜃 were also calculated for the unstratified and stratified Cox PH model fits with serving as the target estimand. Finally, to help understand the key drivers of the main 5-STAR𝜃𝜃 results, we examined the performance of the elastic net filtering step (Step 2) and the CTree step (Step 3) in terms of removal of noise covariates and use of correct (i.e., truly prognostic) covariates to define the risk strata, respectively.

4.2 Results

32

Type I error and power

As shown in Table 2, the type I error rate was well-controlled, on the basis of being less than 0.025 + 2 (0.025 × 0.975)/20,000 = 2.72%, for all the competing methods. In terms of

power, both� the time ratio (TR) and hazard ratio (HR) versions of 5-STAR performed admirably. Under Alt-1, both 5-STAR [TR] and 5-STAR [HR] had 84% power, which was 7 to 17 percentage points higher than the power of the best (stratified logrank) and worst (MaxCombo) performers among the other methods. Under Alt-2, 5-STAR [TR] placed first with 93% power followed by a tie for second place between 5-STAR [HR] and stratified logrank (90% power each), well- separated from the other methods which had powers ranging from 82% to 84%. Finally, under Alt- 3, the best two performers were 5-STAR [HR] with 73% power and 5-STAR [TR] with 67% power, both notably more powerful than the other methods which had powers ranging from 48% to 54%.

Table 2

Summary of simulation results: type I error (target α=2.5%), power, and mean percent bias and coverage of 95% CI for the relevant target estimand (average HR or TR) based on 20,000 simulations under each scenario

Scenario: Null Alt-1 Alt-2 Alt-3 True HRs in risk strata: 1, 1, 1, 1 0.7, 0.7, 0.7, 0.7 0.42, 0.7, 0.86, 0.95 0.95, 0.86, 0.7, 0.42 Type I Power Mean 95% CI Power Mean 95% CI Power Mean 95% CI Analysis Method Error (%) (%) % Bias Coverage (%) % Bias Coverage (%) % Bias Coverage Unstratified logrank/CPH 2.56 71 9.1 88.3 82 4.9 93.3 50 15.9 74.9 Stratified logrank/CPH* 2.49 77 6.1 92.3 90 -0.3 95.0 48 15.9 76.1 MaxCombo 2.60 67 -- -- 83 -- -- 54 -- -- RMST 2.51 71 -- -- 84 -- -- 48 -- -- 5-STAR [TR] 2.49 84 0.3 95.8 93 0.2 95.3 67 0.3 96.0 5-STAR [HR] 2.52 84 0.4 95.9 90 -2.7 94.9 73 1.2 96.1

CPH = Cox proportional hazards model, * analysis strata based on three pre-specified factors (two prognostic and one non-prognostic, in truth), TR = time ratio, HR = hazard ratio

Percent bias and coverage of 95% CI for target estimand

As shown in Table 2, point estimates using 5-STAR [TR] and 5-STAR [HR] were associated with negligible bias for their respective average time ratio (γ) and average hazard ratio (θ) target estimands. Moreover, the 5-STAR 95% confidence intervals captured their true target estimand

33

very close to 95% of the time in every scenario studied. In contrast, the unstratified and stratified Cox PH analyses were associated with point estimates for θ that were notably biased under Alt-1 and Alt-3, with corresponding 95% confidence intervals having considerable under-coverage of the target parameter.

5-STAR: effectiveness of covariate filtering (Step 2) and risk strata formation (Step 3)

The elastic net Cox regression algorithm using pooled survival times blinded to patient-level treatment assignment did a reasonably good job filtering out noise covariates in Step 2 of 5-STAR. Across the four simulated scenarios, on average, approximately 9-10 of the 50 candidate covariates advanced to Step 3, with all of the correct (i.e., truly prognostic) covariates , and

advancing approximately 94-99% of the time. The CTree algorithm used for stratum𝑋𝑋1 formation𝑋𝑋2 𝑋𝑋 26in Step 3 of 5-STAR also performed reasonably well. Across the four simulated scenarios, on average, 3.4 covariates defined the final formed risk strata, with the latter based on at least the three correct covariates 83-98% of the time and based on only the three correct covariates 48-64% of the time.

5. Concluding remarks The power of the ubiquitous logrank test for a between-treatment comparison of survival times in randomized clinical trials can be notably less than desired if the treatment hazard functions are non-proportional, and the accompanying hazard ratio estimate from a Cox proportional hazards model can be hard to interpret. Increasingly popular approaches to guard against the statistical adverse effects of non-proportional hazards include the MaxCombo test (based on a versatile combination of weighted logrank statistics) and a test based on a between-treatment comparison of restricted mean survival time (RMST). Unfortunately, neither the logrank test nor the latter two approaches are designed to leverage what we refer to as structured patient heterogeneity in clinical trial populations, and this can contribute to suboptimal power for detecting a between-treatment difference in the distribution of survival times. Stratified versions of the logrank test and the corresponding Cox proportional hazards model based on pre-specified stratification factors represent steps in the right direction. However, they carry unnecessary risks associated with both

34

a potential suboptimal choice of stratification factors and with potentially implausible dual assumptions of proportional hazards within each stratum and a constant hazard ratio across strata.

We have developed and described a novel alternative to the aforementioned current approaches for survival analysis in randomized clinical trials. Our approach envisions the overall patient population as being a finite mixture of subpopulations (risk strata), with higher to lower ordered risk strata comprised of patients having shorter to longer expected survival regardless of treatment assignment. Patients within a given risk stratum are deemed prognostically homogeneous in that they have in common certain pre-treatment characteristics that jointly strongly associate with survival time. Given this conceptualization and motivated by a reasonable expectation that detection of a true treatment difference should get easier as the patient population gets prognostically more homogeneous, our proposed method follows naturally. Starting with a pre- specified set of baseline covariates (Step 1), elastic net Cox regression (Step 2) and a subsequent conditional inference tree algorithm (Step 3) are used to segment the trial patients into ordered risk strata; importantly, both steps are blinded to patient-level treatment assignment. After unblinding, a treatment comparison is done within each formed risk stratum (Step 4) and stratum-level results are combined for overall estimation and inference (Step 5).

For the primary analysis, labeled 5-STAR [TR], exponentiated estimates of between-treatment differences in mean log survival time, referred to as time ratios, are used for treatment comparisons within the formed risk strata. Estimation is accomplished using three accelerated failure time model fits (assuming survival times follow either a Weibull, log-normal or log-logistic distribution) in conjunction with straightforward model averaging. As a supplemental analysis, labeled 5-STAR [HR], hazard ratio estimates from basic Cox proportional hazard model fits within formed risk strata can be used, with the usual understanding that a hazard ratio estimate is hard to interpret if the corresponding treatment hazard functions are non-proportional. In addition to each formed stratum-level point estimate and 95% confidence interval for a time ratio (and hazard ratio, if needed), we recommend reporting a corresponding estimate of the probability that the test treatment is associated with expected longer survival than the control treatment. This level of detail in stratum-level reporting, currently uncommon in practice, provides transparency that aids in 35

understanding the key inputs for the overall (i.e., stratum-averaged) result. Moreover, it serves as a potential enabler of personalized medicine by drawing attention to any identifiable subgroup of patients defined by the formed strata that may have a notably low likelihood of experiencing longer survival with the test treatment relative to the control treatment despite an overall statistically significant result in favor of the test treatment.

In summary, using a detailed analysis of a hypothetical dataset, retrospective analyses of two real datasets, and results from a simulation study, we have illustrated the impressive power-boosting performance and utility of our proposed 5-step stratified testing and amalgamation routine (5- STAR) relative to that of the logrank test and other common approaches that are not designed to leverage inherently structured patient heterogeneity. We end by making observations on two unrelated but important fronts. First, suppose an interim analysis is planned for a randomized clinical trial to enable a potentially earlier conclusion of either futility or overwhelming success for the test treatment. With application of 5-STAR in mind, it is natural to inquire whether the risk strata formed at the interim analysis should be re-used for the subsequent final analysis (if applicable), or whether the risk strata formation step should be repeated for the latter. Second, even though the focus of this manuscript has been on survival analysis, the 5-STAR concept can be extended to analyses of continuous, ordinal and nominal endpoints. Research on both fronts is ongoing and will be the subject of future communications.

Software: the 5-STAR methodology proposed in this manuscript can be easily implemented using the fiveSTAR R package available at https://github.com/rmarceauwest/fiveSTAR.

REFERENCES 1. Mantel N. Evaluation of survival data and two new rank order statistics arising in its consideration. Cancer Chemother Rep. 1966;50(3):163-170. 2. Peto R, Peto J. Asymptotically efficient rank invariant test procedures. J R Stat Soc Ser A Stat Soc. 1972;135(2):185-207.

36

3. Cox DR. Regression models and life‐tables. J R Stat Soc Series B Stat Methodol. 1972;34(2):187-202. 4. Buntrock C, Ebert DD, Lehr D, et al. Effect of a web-based guided self-help intervention for prevention of major depression in adults with subthreshold depression: a randomized clinical trial. JAMA. 2016;315(17):1854-1863. 5. Chesney J, Puzanov I, Collichio F, et al. Randomized, open-label phase II study evaluating the efficacy and safety of talimogene laherparepvec in combination with ipilimumab versus ipilimumab alone in patients with advanced, unresectable melanoma. J Clin Oncol. 2018;36(17):1658-1667. 6. Tripathy D, Im S-A, Colleoni M, et al. Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): a randomised phase 3 trial. Lancet Oncol. 2018;19(7):904-915. 7. von Minckwitz G, Huang C-S, Mano MS, et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 2019;380(7):617-628. 8. Kaufman PA, Awada A, Twelves C, et al. Phase III open-label randomized study of eribulin mesylate versus capecitabine in patients with locally advanced or metastatic breast cancer previously treated with an anthracycline and a taxane. J Clin Oncol. 2015;33(6):594-601. 9. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non–small-cell lung cancer. N Engl J Med. 2015;373(17):1627-1639. 10. Herbst RS, Baas P, Kim D-W, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540-1550. 11. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255-265. 12. Rulli E, Ghilotti F, Biagioli E, et al. Assessment of proportional hazard assumption in : a systematic review on statistical methodology in clinical trials using time-to-event endpoint. Br J Cancer. 2018;119(12):1456-1463.

37

13. Rahman R, Fell G, Ventz S, et al. Deviation from the proportional hazards assumption in randomized phase 3 clinical trials in oncology: prevalence, associated factors, and implications. Clin Cancer Res. 2019;25(21):6339-6345. 14. Harrington DP, Fleming TR. A class of rank test procedures for censored survival data. Biometrika. 1982;69(3):553-566. 15. Lee JW. Some versatile tests based on the simultaneous use of weighted log-rank statistics. Biometrics. 1996;52(2):721-725. 16. Kosorok MR, Lin C-Y. The versatility of function-indexed weighted log-rank statistics. J Am Stat Assoc. 1999;94(445):320-332. 17. Lee S-H. On the versatility of the combination of the weighted log-rank statistics. Comput Stat Data Anal. 2007;51(12):6557-6564. 18. Kössler W. Max-type rank tests, U-tests, and adaptive tests for the two-sample location problem - an asymptotic power study. Comput Stat Data Anal. 2010;54(9):2053-2065. 19. Garès V, Andrieu S, Dupuy JF, Savy N. An omnibus test for several hazard alternatives in prevention randomized controlled clinical trials. Stat Med. 2015;34(4):541-557. 20. Karrison TG. Versatile tests for comparing survival curves based on weighted log-rank statistics. Stata J. 2016;16(3):678-690. 21. Roychoudhury S, Anderson KM, Ye J, Mukhopadhyay P. Robust design and analysis of clinical trials with non-proportional hazards: a straw man guidance from a cross-pharma working group. arXiv preprint. 2019:arXiv:1908.07112 [stat.AP]. 22. Pepe MS, Fleming TR. Weighted Kaplan-Meier statistics: a class of distance tests for censored survival data. Biometrics. 1989;45(2):497-507. 23. Royston P, Parmar MK. The use of restricted mean survival time to estimate the treatment effect in randomized clinical trials when the proportional hazards assumption is in doubt. Stat Med. 2011;30(19):2409-2421. 24. Tian L, Zhao L, Wei LJ. Predicting the restricted mean event time with the subject's baseline covariates in survival analysis. Biostatistics. 2014;15(2):222-233. 25. Uno H, Claggett B, Tian L, et al. Moving beyond the hazard ratio in quantifying the between-group difference in survival analysis. J Clin Oncol. 2014;32(22):2380-2385. 26. Zhao L, Claggett B, Tian L, et al. On the restricted mean survival time curve in survival analysis. Biometrics. 2016;72(1):215-221.

38

27. Kalbfleisch JD, Prentice RL. The Statistical Analysis of Failure Time Data. 2nd ed. Hoboken, NJ: John Wiley & Sons, Inc.; 2002. 28. Gail MH, Wieand S, Piantadosi S. Biased estimates of treatment effect in randomized with nonlinear regressions and omitted covariates. Biometrika. 1984;71(3):431-444. 29. Anderson GL, Fleming TR. Model misspecification in proportional hazards regression. Biometrika. 1995;82(3):527-541. 30. Mehrotra DV, Su SC, Li X. An efficient alternative to the stratified cox model analysis. Stat Med. 2012;31(17):1849-1856. 31. Ishwaran H, Kogalur UB, Blackstone EH, Lauer MS. Random survival forests. Ann Appl Stat. 2008;2(3):841-860. 32. Ishwaran H, Lu M. Standard errors and confidence intervals for variable importance in random forest regression, classification, and survival. Stat Med. 2019;38(4):558-582. 33. Zou H, Hastie T. Regularization and variable selection via the elastic net. J R Stat Soc Series B Stat Methodol. 2005;67(2):301-320. 34. Park MY, Hastie T. L1‐regularization path algorithm for generalized linear models. J R Stat Soc Series B Stat Methodol. 2007;69(4):659-677. 35. Hoerl AE, Kennard RW. Ridge regression: biased estimation for nonorthogonal problems. Technometrics. 1970;12(1):55-67. 36. Tibshirani R. Regression shrinkage and selection via the lasso. J R Stat Soc Series B Stat Methodol. 1996;58(1):267-288. 37. Hothorn T, Hornik K, Zeileis A. Unbiased recursive partitioning: a conditional inference framework. J Comput Graph Stat. 2006;15(3):651-674. 38. Šidák Z. Rectangular confidence regions for the means of multivariate normal distributions. J Am Stat Assoc. 1967;62(318):626-633. 39. Chiou SH, Kang S, Yan J. Fitting accelerated failure time model in routine survival analysis with R package aftgee. J Stat Softw. 2014;61(11):1-23. 40. Liao JJZ, Liu GF. A flexible parametric survival model for fitting time to event data in clinical trials. Pharm Stat. 2019;18(5):555-567. 41. Buckland ST, Burnham KP, Augustin NH. : an integral part of inference. Biometrics. 1997;53(2):603-618.

39

42. Nadarajah S, Kotz S. Exact distribution of the max/min of two Gaussian random variables. IEEE Trans Very Large Scale Integr VLSI Syst. 2008;16(2):210-212. 43. Permutt T. Do Covariates Change the Estimand? Stat Biopharm Res. 2020;12(1):45-53. 44. Grambsch PM, Therneau TM. Proportional hazards tests and diagnostics based on weighted residuals. Biometrika. 1994;81(3):515-526. 45. Lipkovich I, Dmitrienko A, D'Agostino Sr RB. Tutorial in biostatistics: data‐driven subgroup identification and analysis in clinical trials. Stat Med. 2017;36(1):136-196. 46. Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372(25):2387-2397.

40